US10174286B2 - Pancreatic islet-like cell structures and a method of preparing thereof - Google Patents

Pancreatic islet-like cell structures and a method of preparing thereof Download PDF

Info

Publication number
US10174286B2
US10174286B2 US15/103,317 US201415103317A US10174286B2 US 10174286 B2 US10174286 B2 US 10174286B2 US 201415103317 A US201415103317 A US 201415103317A US 10174286 B2 US10174286 B2 US 10174286B2
Authority
US
United States
Prior art keywords
cells
cell
poly
structures
islets
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US15/103,317
Other versions
US20160312189A1 (en
Inventor
Ciro Tetta
Giovanni Camussi
Sara Giunti
Victor Manuel NAVARRO TABLEROS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
PRESENIUS MEDICAL CARE DEUTSCHLAND GmbH
Fresenius Medical Care Deutschland GmbH
Original Assignee
PRESENIUS MEDICAL CARE DEUTSCHLAND GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by PRESENIUS MEDICAL CARE DEUTSCHLAND GmbH filed Critical PRESENIUS MEDICAL CARE DEUTSCHLAND GmbH
Assigned to FRESENIUS MEDICAL CARE DEUTSCHLAND GMBH reassignment FRESENIUS MEDICAL CARE DEUTSCHLAND GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TETTA, CIRO, CAMUSSI, GIOVANNI, GIUNTI, Sara, NAVARRO TABLEROS, Victor Manuel
Publication of US20160312189A1 publication Critical patent/US20160312189A1/en
Application granted granted Critical
Publication of US10174286B2 publication Critical patent/US10174286B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0676Pancreatic cells
    • C12N5/0677Three-dimensional culture, tissue culture or organ culture; Encapsulated cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/507Pancreatic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/14Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from hepatocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers
    • C12N2533/32Polylysine, polyornithine

Definitions

  • the present invention relates to the field stem cell differentiation.
  • the present invention relates to a method for differentiating a stem cell into a pancreatic islet-like cell structure and to the pancreatic islet-like cell structure obtained therefrom.
  • Type 1 diabetes is a disease with a huge socio-economic impact that, if untreated, leads to death. It is caused by autoimmune destruction of pancreatic insulin producing cells ( ⁇ -cells) and associated with the development of debilitating microvascular and macrovascular complications.
  • ⁇ -cells pancreatic insulin producing cells
  • stem cells represent a promising source for the generation of insulin-producing cells, as they may have properties that allow to satisfy the requirements for the cure of type 1 diabetes mellitus (T1DM). Firstly, they may allow both to restore ⁇ -cell function and prevent recurrence of autoimmunity, without the need of using immunosuppressive therapy. Secondly, due to their potentially unlimited capacity for self-renewal, they could be broadly used.
  • T1DM type 1 diabetes mellitus
  • stem cells isolated from human amniotic fluid and dental pulp (Carnevale G et al. Dig Liver Dis. 2013; 45(8): 669-76), and human umbilical cord blood-derived mesenchymal stem cells (Prabakar K R et al. Cell Transplant. 2012; 21(6): 1321-39) are disclosed in the prior art.
  • islets-like structures derived in vitro from human embryonic stem cells were found to be immature, thus requiring a further in vivo 3-to-4 month differentiation period in order to become mature functional islets (Kroon E et al. Nat Biotechnol. 2008; 26(4): 443-52; Rezania A et al. Diabetes. 2012; 61(8): 2016-29).
  • Insulin-producing cells generated in vitro from human adipose tissue-derived stem cells (Mohamad Buang M L et al. Arch Med Res. 2012; 43(1): 83-8; Chandra V et al. PLoS One. 2011; 6(6):e20615), nestin-positive cells derived from bone marrow (Milanesi A et al. J Endocrinol. 2011; 209(2); 193-201), stem cells isolated from human amniotic fluid and dental pulp (Carnevale G et al. Dig Liver Dis. 2013; 45(8): 669-76) were shown to release human insulin in response to glucose stimulation in vitro. However, expression of glucagon, PP and ghrelin was not demonstrated.
  • CB-MSC human umbilical cord blood-derived mesenchymal stem cells
  • HLSCs human liver stem cells
  • HLSCs were shown to change their elongated morphology into small spheroid cell clusters, morphologically resembling pancreatic islets, when cultured in DMEM with high glucose content (23 mM) for a month followed by 5-7 days of culture in the presence of 10 mM nicotinamide.
  • DMEM high glucose content
  • the differentiation protocol was limited by a low efficiency, particularly referring to both the quantity of the generated structures and the time required for generating them. Furthermore, even after extensive culturing the structures did not quite grow to a size and morphology closely resembling human pancreatic structures.
  • pancreatic islet-like cell structures which, advantageously, have a diameter comparable to the naturally-occurring human pancreatic islets and which express, at the protein level, all of the hormones which are usually produced by human pancreatic islets (i.e. insulin, glucagon, PP, somatostatin and ghrelin).
  • the pancreatic islet-like cell structures obtainable by the method of the present invention are capable of reducing blood glucose levels significantly early (within 13 days), with a concomitant increase in human C-peptide.
  • the pancreatic islet-like cell structures generated with the method of the invention render unique the pancreatic islet-like cell structures generated with the method of the invention.
  • no previous studies have shown such a combination of in vitro and in vivo features in islet-like structures derived from a single isolated stem cell type. Due to their features, the pancreatic islet-like cell structures obtainable by the method of the present invention are particularly suitable for use in various applications, such as basic research, drug screening and regenerative medicine.
  • the invention provides a method of preparing an artificially grown spheroid pancreatic islet-like cell structure, which comprises the step of culturing an isolated adult stem cell in a first differentiation liquid cell culture medium which comprises a poly-cationic substance.
  • isolated adult stem cells when cultured in a liquid cell culture medium in the presence of a poly-cationic substance, quite early (i.e. after about 2 to 4 days) aggregate and differentiate into spheroid pancreatic islet-like cell structures, which closely resemble naturally-occurring pancreatic islet cell structures both in size and morphology, and which are capable of expressing the hormones which are usually produced by human pancreatic islets (i.e. insulin, glucagon, PP, somatostatin and ghrelin).
  • a poly-cationic substance quite early (i.e. after about 2 to 4 days) aggregate and differentiate into spheroid pancreatic islet-like cell structures, which closely resemble naturally-occurring pancreatic islet cell structures both in size and morphology, and which are capable of expressing the hormones which are usually produced by human pancreatic islets (i.e. insulin, glucagon, PP, somatostatin and ghrelin).
  • the adult stem cell used in the method of the invention is preferably an adult mammalian stem cell. Therefore, any liquid cell culture medium capable of sustaining the growth of mammalian cells is suitable for use in the method of the invention.
  • the liquid cell culture medium is a serum-enriched cell culture medium. Serum-enriched DMEM or RPMI are mentioned as non-limiting examples. Serum concentration in the cell culture medium is preferably comprised within the range of from 5 to 20%.
  • the liquid cell culture medium also comprises one or more carbon sources, such as for example glucose and glutamine. Preferred glucose concentrations are within the range of 6-25 mM. Preferred glutamine concentrations are within the range of 0.5-3 mM.
  • adult stem cell is intended to mean a stem cell that is isolated from an adult tissue, in contrast with an “embryonic stem cell” which is isolated from the inner cell mass of a blastocyst.
  • embryonic stem cell which is isolated from the inner cell mass of a blastocyst.
  • embryonic stem cells are also known as “somatic stem cells”.
  • Any poly-cationic substance which is capable of promoting adult stem cell aggregation and differentiation into pancreatic islet cells at a given concentration and which, at that concentration is non-cytotoxic to cells, may be used in the method of the present invention.
  • poly-cationic substances suitable for use in the method of the invention are poly-lysine and protamine.
  • Protamine which is the preferred poly-cationic substance because it is suitable for clinical applications, is preferably added to the medium in the form of a soluble salt, such as for example protamine sulfate or protamine hydrochloride, at a concentration which preferably ranges between 5 and 20 mM.
  • a soluble salt such as for example protamine sulfate or protamine hydrochloride
  • the method of the invention further comprises the step of replacing the first differentiation liquid cell culture medium with a second differentiation liquid cell culture medium not comprising a poly-cationic substance and culturing the cells in said second medium.
  • a second differentiation liquid cell culture medium not comprising a poly-cationic substance
  • Any liquid cell culture medium capable of sustaining the growth of mammalian cells is suitable for use as the second differentiation liquid cell culture medium.
  • the same liquid cell culture medium which was used in the first step shall also be employed in the second step, except that it does not contain the poly-cationic substance.
  • the aim of this step is to obtain complete maturation of the pancreatic islet-like cell structures formed during the first step as well as their increase both in cell number and in structure number.
  • Cultivation in the second differentiation liquid cell culture medium is preferably carried out for a period of time of at least 2 days, more preferably for at least 10 days, even more preferably for about 10 to 14 days.
  • the presence of a poly-cationic substance in the first differentiation liquid cell culture medium is the key element of the invention, in that it accelerates the formation of spheroid cell clusters resembling islets-like structures which closely resemble naturally occurring human pancreatic islet structures.
  • HLSCs exposure to either DMEM or RPMI-based differentiation media did not result in islet-like structures formation after a culture period of 4 days.
  • the addition of protamine to either a DMEM or RPMI-based medium both in the presence and in the absence of glucose, resulted in islet-like structures formation after a culture period of 4 days ( FIG. 11 ).
  • Diameter of the structures derived from HLSCs is comparable to that reported for human pancreatic islets (Chandra V et al. PLoS One. 2011; 6(6):e20615) and mostly comprized between 50 and 150 um, with an average islets volume of 109 ⁇ 19 uM (mean ⁇ SD) ( FIG. 12 ).
  • glucose did not affect islet-like structures formation ( FIG. 13A ), but it plays a key role in both inducing endocrine specification (NgN3 expression) and increasing both insulin and glucagon expression within the structures ( FIG. 13B ).
  • the adult stem cell which is differentiated into pancreatic islet-like cell structures is an adult human liver stem cell (HLSC).
  • a preferred human liver stem cell is the human non-oval liver stem cell (HLSC) expressing both mesenchymal and embryonic stem cell markers disclosed in WO 2006/126219.
  • This cell line is in particular characterized in that it is a non-oval human liver pluripotent progenitor cell line which is isolated from adult tissue, which expresses hepatic cell markers and which has multipotent differentiation abilities and regenerative properties. More in particular, this cell line is capable of differentiating into mature liver cells, insulin producing cells, osteogenic cells and epithelial cells.
  • it expresses one or more markers selected from the group comprising albumin, ⁇ -fetoprotein, CK18, CD44, CD29, CD73, CD146, CD105, CD90 and any combination thereof, and it does not express markers selected from the group comprising CD133, CD117, CK19, CD34, cytochrome P450.
  • the human non-oval liver pluripotent progenitor/stem cells disclosed in WO 2006/126236 were shown to undergo differentiation into a variety of tissue cell types (namely, mature liver cells, epithelial cells, insulin-producing cells and osteogenic cells) and to exert organ regenerating effects.
  • tissue cell types namely, mature liver cells, epithelial cells, insulin-producing cells and osteogenic cells
  • Such cells are derived from a non-oval human liver pluripotent progenitor cell line which expresses hepatic cell markers.
  • Such cells are isolated by a method comprising the steps of:
  • HLSCs in the present invention are preferred for a number of reasons: 1) they are relatively easy to isolate and expand, 2) they are characterized by a degree of proliferation that allows to provide a suitable number of cells for therapeutic use, 3) they can be used autologously and 4) they avoid ethical concerns. Furthermore, the liver and the pancreas share common embryonic origins and the developing liver has been shown to exhibit transcriptional features similar to the adult pancreas (Bose B et al. Cell Biol Int. 2012; 36(11): 1013-20).
  • poly-cationic substances shall be effective in promoting aggregation and differentiation of other adult stem cells than HLSCs
  • the scope of the invention is not limited to the use of HLSCs only, but it includes the use of any adult stem cells type.
  • pancreatic islet-like cells structures obtained by the method of the present invention are characterized by a unique combination of morphological and functional features which is not observed in other islet-like cells structures prepared by the methods of the prior art. Accordingly, the scope of the invention also includes a spheroid pancreatic islet-like cell structure as defined in the appended claims.
  • the spheroid pancreatic islet-like cell structure according to the present invention is advantageously characterized by a diameter of from 50 to 250 ⁇ m, preferably of from 50 to 200 ⁇ m, which is comparable to the naturally-occurring human pancreatic islets. It is also characterized by a volume, expressed as IEQ/100 ILS, ranging from 30 to 200 ⁇ m, preferably from 50 to 130 ⁇ m and, even more advantageously, by the ability to express, at the protein level, all of the pancreatic hormones which are usually expressed by human pancreatic islets, i.e. insulin, glucagon, pancreatic polypeptide, somatostatin and ghrelin.
  • IEQ/100 ILS is the Islets Equivalent of an average diameter of 150 ⁇ m (IEQ) normalized to 100 islets (ILS).
  • IEQ Islets Equivalent of an average diameter of 150 ⁇ m
  • ILS islets normalized to 100 islets
  • IEQ of islets isolated from the pancreas and used for transplantation is determined as follows. Suspended islets isolated from the pancreas are evaluated both in number and size (diameter) in order to determine the total islets equivalent (i.e. total islets volume). Islets diameter assessment takes into account 50 ⁇ m diameter range increments, from 50 am to >350 ⁇ m, without taking into account diameters ⁇ 50 ⁇ m, that do not provide a significant contribution to the total volume.
  • a relative conversion factor is conventionally used to convert the total islets number to Islets Equivalent of an average diameter of 150 ⁇ m (IEQ).
  • Suspended islets isolated from the pancreas are separated in different layers based on their degree of purity.
  • An islet sample from each layer of the final product is then analyzed: the total number of islets and the IEQ are then calculated based on the number, the purity and the size of the islets, using standardized methods.
  • the islet-like structures are obtained from cultured cells, that are adherent to the plate, the inventors assessed both the number and the diameter of the islet-like structures by analyzing 200 10 ⁇ micrographs, randomly selected, per experiment, and expressed the results as % ⁇ SD ( FIG. 12 A) and IEQ normalized to 100 islets ( FIG. 12 C).
  • the spheroid pancreatic islet-like cell structure according to the present invention are particularly suitable for use both in clinical application, such as in a method treatment of diabetes by pancreatic islet transplantation, and in in vitro applications, such as screening methods for identifying substances capable of promoting the expression of one or more pancreatic hormones by pancreatic islet cells or for identifying substances capable of exerting a cytotoxic effect on pancreatic islet cells.
  • HLSCs lines are isolated from healthy liver tissues of patients undergoing hepatectomies and characterized as previously described (Mohamad Buang M L et al. Arch Med Res. 2012; 43(1): 83-8). Specifically, HLSCs are seeded in 75 cm 3 culture flasks and cultured in a medium (see Table 1) containing a 3 to 1 proportion of ⁇ -minimum essential medium and endothelial cell basal medium-1, supplemented with L-glutamine 2 mM, penicillin 100 UI/ml/streptomycin 100 ⁇ g/ml and 10% Fetal Bovine Serum. Cells are maintained in a humidified 5% CO 2 incubator at 37° C.
  • cells are washed twice with PBS and incubated with trypsin-EDTA 1 ⁇ for about 5 minutes at 37° C., in order to induce cell detachment. Trypsin activity is subsequently neutralized by adding RPMI supplemented with L-glutamine 2 mM, penicillin 100 UI/ml/streptomycin 100 ⁇ g/ml and 10% Fetal Bovine Serum. Then, cells are harvested by centrifugation at 1200 rpm for 5 minutes, the supernatant is removed and the pellet re-suspended in culture medium and split among five 75 cm 3 culture flasks.
  • Cells up to a ⁇ 80% confluence are detached and harvested by centrifugation as described in paragraph 1.2.
  • the cells are counted and 10 6 cells per vial are cryopreserved.
  • the cell pellet is resuspended in a 1 ml solution containing 90% FBS and 10% dimethyl sulphoxide (DMSO) and placed into pre-cooled cryovial/s.
  • the cryovials are frozen at ⁇ 80° C. overnight before being placed into the liquid nitrogen container at ⁇ 196° C.
  • a vial of frozen HLSCs is removed from the liquid nitrogen tank and placed in a beaker of water pre-warmed to 37° C. Once the cell suspension has completely thawed, it is placed into a sterile 50 ml falcon tube with 10 ml of sterile media and centrifuged at 1200 rpm for 5 minutes. The cell pellet is then resuspended in culture medium and split in three 75 cm 3 culture flask and left to attach overnight at 37° C. in a humidified 5% CO 2 incubator. The medium is changed the following day.
  • HLSCs at a density of 12 ⁇ 10 3 /cm 2 are seeded in 25 cm 3 culture flasks or in a Petri dish 100 ⁇ 20 mm in the differentiation culture medium 1 (see Table 1) consisting in RPMI 1640 or DMEM supplemented with 10% Fetal Bovine Serum, glucose 11.6 mM, protamin chloride 10 ⁇ g/ml, L-glutamine 2 mM and penicillin 100 UI/ml/streptomycin 100 ⁇ g/ml.
  • Cells are placed, for a period of 4 days, without changing the medium, in a humidified 5% CO 2 incubator at 37° C.
  • differentiation culture medium 2 consisting in RPMI 1640 or DMEM supplemented with 10% FBS, glucose 11.6 mM, L-glutamine 2 mM and penicillin 100 UI/ml/streptomycin 100 ⁇ g/ml.
  • Medium is subsequently changed every other day.
  • cells are expected to start organizing in islets-like structures that reach a maximum number after 14-18 days ( FIG. 1 ).
  • HLSC differentiation medium 1 Final concentration Volume (ml) Final volume 250 RPMI 1640 or DMEM 216.85 FBS 10% 25 Penicillin/streptomycin 100 UI/ml/100 ug/ml 2.5 Glutamine 200 mM 2 mM 2.5 Glucose 1M 11.6 mM 2.9 Protamine 10 mg/ml 10 ug/ml 0.25
  • HLSC differentiation medium 2 Final concentration Volume (ml) Final volume 250 RPMI or DMEM 217.1 FBS 10% 25 Penicillin/streptomycin 100 UI/ml/100 ug/ml 2.5 Glutamine 200 mM 2 mM 2.5 Glucose 1M 11.6 mM 2.9
  • FIG. 1 representative 20 ⁇ micrographs showing HLSCs in basal culture medium (A) and following culture in differentiation medium with protamine (B-D). After 24 h, the cells changed in morphology and started forming small clusters (B) that progressively increased in both size and number, reaching a maximum number after a culture period of 18 days (C).
  • B 40 ⁇ micrograph showing islets-like structures after a culture period of 18 days.
  • E Graph showing the total number of islets-like structures per 25 cm 3 culture flask after 18 days of culture.
  • FIGS. 2-8 representative pictures showing islet-like structures characterization by immunofluorescence after 14 days of culture: islets-like structures become positively stained for both PDX-1 and NgN3 ( FIG. 2 ), insulin and glucagon ( FIG. 3 ), C-peptide and GLUT-2 ( FIG. 4 ), somatostatin ( FIG. 5 ), ghrelin and PP ( FIG. 6 ), collagen IV ( FIG. 7 ) and von Willebrand Factor ( FIG. 8 ).
  • FIG. 9 representative pictures showing immunofluorescence characterization of islet-like structures cells following islet-like structures disaggregation (trypsin 1 ⁇ ) after 14 days of culture.
  • FIG. 10 graph showing both blood glucose and human C-peptide levels in non-diabetic SCID mice (solid line, CTRL), streptozotocin-induced (55 mg/kg/day for 5 days) diabetic SCID mice (dotted line, DM) and streptozotocin-induced diabetic SCID mice that received HLSC derived islets-like structures implant (5000 IEQ/kg) under the renal capsule (dashed line, DM+ILS), before and 13 days following the implant.
  • HLSC derived islets-like structures implant 5000 IEQ/kg under the renal capsule (dashed line, DM+ILS), before and 13 days following the implant.
  • diabetic mice that received islets-like structures implant had a significant decrease in blood glucose levels. This was paralleled by a concomitant increase in human C-peptide, that remained undetectable in both non diabetic SCID mice and in diabetic SCID mice that did not receive the implant.
  • FIG. 11 graph showing islets-like structure (ILS) formation following HLSCs culture in RPMI or DMEM based medium supplemented with FBS 10% (F), FBS 10%+ glucose 11.6 mM (FG) or FBS 10%+ glucose 11.6 mM+ protamine chloride 10 ⁇ g/ml (FGP) for 48 hours.
  • FIG. 12 graphs showing the size distribution (A), the mean ⁇ SD diameter (B) and IEQ/100 ILS (C) of islets-like structures derived from HLSCs following 14 days of culture in RPMI based medium supplemented with FBS 10%+ glucose 11.6 mM+ protamine chloride 10 ⁇ g/ml.
  • FIG. 13 Glucose does not affect islets-like structures (ILS) formation (A), but plays a key role in inducing endocrine specification and insulin/glucagon expression (B).
  • B Representative picture showing PDX-1, NgN3, insulin and glucagon expression in RPMI-based medium with glucose 1 or 25 mM.
  • FIG. 14 representative pictures showing the expression of PDX-1, NgN3, GLUT-2, C-peptide, glucagon and somatostatin in cells cultivated in a poly-lysine differentiation medium.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Transplantation (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Botany (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Toxicology (AREA)
  • Analytical Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Endocrinology (AREA)

Abstract

The invention relates to a method of preparing pancreatic islet-like cell structures characterized by a unique combination of morphological and functional features which make them particularly suitable for use in both clinical and drug screening application, as well as to the pancreatic islet-like cell structures obtained therefrom.

Description

The present invention relates to the field stem cell differentiation. In particular, the present invention relates to a method for differentiating a stem cell into a pancreatic islet-like cell structure and to the pancreatic islet-like cell structure obtained therefrom.
Type 1 diabetes is a disease with a huge socio-economic impact that, if untreated, leads to death. It is caused by autoimmune destruction of pancreatic insulin producing cells (β-cells) and associated with the development of debilitating microvascular and macrovascular complications.
Both pancreas transplantation and islets transplantation have been shown to restore islets function and potentially reduce long-term diabetic complications, but are limited by both donor shortage and the need for immunosuppression. In this context, stem cells represent a promising source for the generation of insulin-producing cells, as they may have properties that allow to satisfy the requirements for the cure of type 1 diabetes mellitus (T1DM). Firstly, they may allow both to restore β-cell function and prevent recurrence of autoimmunity, without the need of using immunosuppressive therapy. Secondly, due to their potentially unlimited capacity for self-renewal, they could be broadly used.
In vitro methods to generate, under specific culture conditions, islets-like structures from human embryonic stem cells (Kroon E et al. Nat Biotechnol. 2008; 26(4): 443-52; Rezania A et al. Diabetes. 2012; 61(8): 2016-29; Bose B et al. Cell Biol Int. 2012; 36(11): 1013-20), human adipose tissue-derived stem cells (Mohamad Buang M L et al. Arch Med Res. 2012; 43(1): 83-8; Chandra V et al. PLoS One. 2011; 6(6):e20615), nestin-positive cells derived from bone marrow (Milanesi A et al. J Endocrinol. 2011; 209(2); 193-201), stem cells isolated from human amniotic fluid and dental pulp (Carnevale G et al. Dig Liver Dis. 2013; 45(8): 669-76), and human umbilical cord blood-derived mesenchymal stem cells (Prabakar K R et al. Cell Transplant. 2012; 21(6): 1321-39) are disclosed in the prior art.
However, islets-like structures derived in vitro from human embryonic stem cells were found to be immature, thus requiring a further in vivo 3-to-4 month differentiation period in order to become mature functional islets (Kroon E et al. Nat Biotechnol. 2008; 26(4): 443-52; Rezania A et al. Diabetes. 2012; 61(8): 2016-29). Furthermore, one study reporting 3D in vitro generation of insulin secreting cells from human embryonic stem cells, while showing a stable trend to a decrease in glycaemia from 300 mg/dl to 200 mg/dl, did not demonstrate, with the exception of insulin, protein expression of other hormones usually produced within the pancreatic islets (glucagon, pancreatic polypeptide (PP), somatostatin and ghrelin) (Bose B et al. Cell Biol Int. 2012; 36(11): 1013-20).
Insulin-producing cells generated in vitro from human adipose tissue-derived stem cells (Mohamad Buang M L et al. Arch Med Res. 2012; 43(1): 83-8; Chandra V et al. PLoS One. 2011; 6(6):e20615), nestin-positive cells derived from bone marrow (Milanesi A et al. J Endocrinol. 2011; 209(2); 193-201), stem cells isolated from human amniotic fluid and dental pulp (Carnevale G et al. Dig Liver Dis. 2013; 45(8): 669-76) were shown to release human insulin in response to glucose stimulation in vitro. However, expression of glucagon, PP and ghrelin was not demonstrated.
Insulin-producing cells generated from human umbilical cord blood-derived mesenchymal stem cells (CB-MSC) were shown to express insulin, C-peptide, glucagon and pancreatic polypeptide and to release C-peptide in response to glucose stimulation in vitro. However, the ability of such cells to reduce blood glucose in experimental diabetes was not demonstrated, as the authors only showed in vivo differentiation of engrafted CB-MSC-derived pancreatic endodermal cells into functional endocrine cells with detection of low levels of C-peptide following glucose challenge 60 days after the implantation (Prabakar K R et al. Cell Transplant. 2012; 21(6): 1321-39).
Methods of differentiation of human liver stem cells (HLSCs) into pancreatic islet cells are disclosed in Herrera M B et al. Stem Cells. 2006; 24(12):2840-50 and in International patent application WO 2006/126236.
In Herrera M B et al. Stem Cells. 2006; 24(12):2840-50, HLSCs were shown to change their elongated morphology into small spheroid cell clusters, morphologically resembling pancreatic islets, when cultured in DMEM with high glucose content (23 mM) for a month followed by 5-7 days of culture in the presence of 10 mM nicotinamide. Such spheroid cell clusters were positively stained for human insulin and Glut2 and became positively stained with the zinc-chelating agent dithizone, which is specific for insulin-containing granules, suggesting the differentiation of HLSCs into islet-like structures. Despite such exciting results, the differentiation protocol was limited by a low efficiency, particularly referring to both the quantity of the generated structures and the time required for generating them. Furthermore, even after extensive culturing the structures did not quite grow to a size and morphology closely resembling human pancreatic structures.
In order to overcome the drawbacks of the prior art, the present inventors have provided a new method of preparing a spheroid pancreatic islet-like cell structure, which is defined in the appended claims. The content of the claims forms an integral part of the description.
Compared to the prior art, not only the method according to the present invention is simpler, faster and more efficient, but it also results in structures which more closely resemble natural human pancreatic islet structures both in size and morphology. It results in the generation of a higher number of pancreatic islet-like cell structures which, advantageously, have a diameter comparable to the naturally-occurring human pancreatic islets and which express, at the protein level, all of the hormones which are usually produced by human pancreatic islets (i.e. insulin, glucagon, PP, somatostatin and ghrelin). Moreover, following implant under the kidney capsule of streptozotocin-induced diabetic SCID mice, the pancreatic islet-like cell structures obtainable by the method of the present invention are capable of reducing blood glucose levels significantly early (within 13 days), with a concomitant increase in human C-peptide. To the inventors' knowledge, such strong similarity of structural features to natural human pancreatic islet structures render unique the pancreatic islet-like cell structures generated with the method of the invention. Indeed, no previous studies have shown such a combination of in vitro and in vivo features in islet-like structures derived from a single isolated stem cell type. Due to their features, the pancreatic islet-like cell structures obtainable by the method of the present invention are particularly suitable for use in various applications, such as basic research, drug screening and regenerative medicine.
Thus, in a first aspect, the invention provides a method of preparing an artificially grown spheroid pancreatic islet-like cell structure, which comprises the step of culturing an isolated adult stem cell in a first differentiation liquid cell culture medium which comprises a poly-cationic substance.
Actually, the inventors surprisingly found that isolated adult stem cells, when cultured in a liquid cell culture medium in the presence of a poly-cationic substance, quite early (i.e. after about 2 to 4 days) aggregate and differentiate into spheroid pancreatic islet-like cell structures, which closely resemble naturally-occurring pancreatic islet cell structures both in size and morphology, and which are capable of expressing the hormones which are usually produced by human pancreatic islets (i.e. insulin, glucagon, PP, somatostatin and ghrelin).
The adult stem cell used in the method of the invention is preferably an adult mammalian stem cell. Therefore, any liquid cell culture medium capable of sustaining the growth of mammalian cells is suitable for use in the method of the invention. In a preferred embodiment, the liquid cell culture medium is a serum-enriched cell culture medium. Serum-enriched DMEM or RPMI are mentioned as non-limiting examples. Serum concentration in the cell culture medium is preferably comprised within the range of from 5 to 20%. Preferably, the liquid cell culture medium also comprises one or more carbon sources, such as for example glucose and glutamine. Preferred glucose concentrations are within the range of 6-25 mM. Preferred glutamine concentrations are within the range of 0.5-3 mM.
Within the context of the present description, the expression “adult stem cell” is intended to mean a stem cell that is isolated from an adult tissue, in contrast with an “embryonic stem cell” which is isolated from the inner cell mass of a blastocyst. Adult stem cells are also known as “somatic stem cells”.
Any poly-cationic substance which is capable of promoting adult stem cell aggregation and differentiation into pancreatic islet cells at a given concentration and which, at that concentration is non-cytotoxic to cells, may be used in the method of the present invention.
Illustrative, non-limiting examples of poly-cationic substances suitable for use in the method of the invention are poly-lysine and protamine.
Protamine, which is the preferred poly-cationic substance because it is suitable for clinical applications, is preferably added to the medium in the form of a soluble salt, such as for example protamine sulfate or protamine hydrochloride, at a concentration which preferably ranges between 5 and 20 mM.
In another preferred embodiment, the method of the invention further comprises the step of replacing the first differentiation liquid cell culture medium with a second differentiation liquid cell culture medium not comprising a poly-cationic substance and culturing the cells in said second medium. Any liquid cell culture medium capable of sustaining the growth of mammalian cells is suitable for use as the second differentiation liquid cell culture medium. According to a preferred embodiment, the same liquid cell culture medium which was used in the first step shall also be employed in the second step, except that it does not contain the poly-cationic substance. The aim of this step is to obtain complete maturation of the pancreatic islet-like cell structures formed during the first step as well as their increase both in cell number and in structure number. Cultivation in the second differentiation liquid cell culture medium is preferably carried out for a period of time of at least 2 days, more preferably for at least 10 days, even more preferably for about 10 to 14 days.
As mentioned above, the presence of a poly-cationic substance in the first differentiation liquid cell culture medium is the key element of the invention, in that it accelerates the formation of spheroid cell clusters resembling islets-like structures which closely resemble naturally occurring human pancreatic islet structures.
Indeed, as illustrated in the examples which follow, HLSCs exposure to either DMEM or RPMI-based differentiation media (see Tables 2 and 3 below), both in the presence and in the absence of glucose, did not result in islet-like structures formation after a culture period of 4 days. By contrast, the addition of protamine to either a DMEM or RPMI-based medium, both in the presence and in the absence of glucose, resulted in islet-like structures formation after a culture period of 4 days (FIG. 11). Diameter of the structures derived from HLSCs is comparable to that reported for human pancreatic islets (Chandra V et al. PLoS One. 2011; 6(6):e20615) and mostly comprized between 50 and 150 um, with an average islets volume of 109±19 uM (mean±SD) (FIG. 12).
In contrast, glucose did not affect islet-like structures formation (FIG. 13A), but it plays a key role in both inducing endocrine specification (NgN3 expression) and increasing both insulin and glucagon expression within the structures (FIG. 13B).
According to another preferred embodiment of the invention, the adult stem cell which is differentiated into pancreatic islet-like cell structures is an adult human liver stem cell (HLSC). A preferred human liver stem cell is the human non-oval liver stem cell (HLSC) expressing both mesenchymal and embryonic stem cell markers disclosed in WO 2006/126219. This cell line is in particular characterized in that it is a non-oval human liver pluripotent progenitor cell line which is isolated from adult tissue, which expresses hepatic cell markers and which has multipotent differentiation abilities and regenerative properties. More in particular, this cell line is capable of differentiating into mature liver cells, insulin producing cells, osteogenic cells and epithelial cells. According to a preferred embodiment, it expresses one or more markers selected from the group comprising albumin, α-fetoprotein, CK18, CD44, CD29, CD73, CD146, CD105, CD90 and any combination thereof, and it does not express markers selected from the group comprising CD133, CD117, CK19, CD34, cytochrome P450.
The human non-oval liver pluripotent progenitor/stem cells disclosed in WO 2006/126236 were shown to undergo differentiation into a variety of tissue cell types (namely, mature liver cells, epithelial cells, insulin-producing cells and osteogenic cells) and to exert organ regenerating effects. Such cells are derived from a non-oval human liver pluripotent progenitor cell line which expresses hepatic cell markers. Such cells are isolated by a method comprising the steps of:
    • (i) culturing adult liver-derived human mature hepatocytes in a cell culture medium until death of mature hepatocytes and selection of a population of surviving cells having epithelioid morphology;
    • (ii) expanding the population of surviving cells having epithelioid morphology by culturing in a serum-containing, glucose-containing culture medium supplemented with hEGF (human epithelial growth factor) and bFGF (basic fibroblast growth factor) and comprising the usual inorganic salts, amino acids and vitamins necessary for the growth of mammalian cells
    • and in particular wherein the mature hepatocytes are frozen in a serum-containing culture medium in the presence of a cryoprotecting agent and then thawed prior to culturing according to step (i).
The characterization of the human non-oval liver stem/progenitor cells disclosed in WO 2006/126236 and the method of preparing thereof are herein fully incorporated by reference.
The use of HLSCs in the present invention is preferred for a number of reasons: 1) they are relatively easy to isolate and expand, 2) they are characterized by a degree of proliferation that allows to provide a suitable number of cells for therapeutic use, 3) they can be used autologously and 4) they avoid ethical concerns. Furthermore, the liver and the pancreas share common embryonic origins and the developing liver has been shown to exhibit transcriptional features similar to the adult pancreas (Bose B et al. Cell Biol Int. 2012; 36(11): 1013-20). However, since it is envisaged that poly-cationic substances shall be effective in promoting aggregation and differentiation of other adult stem cells than HLSCs, the scope of the invention is not limited to the use of HLSCs only, but it includes the use of any adult stem cells type.
As mentioned above, the pancreatic islet-like cells structures obtained by the method of the present invention, are characterized by a unique combination of morphological and functional features which is not observed in other islet-like cells structures prepared by the methods of the prior art. Accordingly, the scope of the invention also includes a spheroid pancreatic islet-like cell structure as defined in the appended claims.
In particular, the spheroid pancreatic islet-like cell structure according to the present invention is advantageously characterized by a diameter of from 50 to 250 μm, preferably of from 50 to 200 μm, which is comparable to the naturally-occurring human pancreatic islets. It is also characterized by a volume, expressed as IEQ/100 ILS, ranging from 30 to 200 μm, preferably from 50 to 130 μm and, even more advantageously, by the ability to express, at the protein level, all of the pancreatic hormones which are usually expressed by human pancreatic islets, i.e. insulin, glucagon, pancreatic polypeptide, somatostatin and ghrelin.
Within the context of the present description, IEQ/100 ILS is the Islets Equivalent of an average diameter of 150 μm (IEQ) normalized to 100 islets (ILS). According to conventional protocols, the IEQ of islets isolated from the pancreas and used for transplantation is determined as follows. Suspended islets isolated from the pancreas are evaluated both in number and size (diameter) in order to determine the total islets equivalent (i.e. total islets volume). Islets diameter assessment takes into account 50 μm diameter range increments, from 50 am to >350 μm, without taking into account diameters <50 μm, that do not provide a significant contribution to the total volume. A relative conversion factor is conventionally used to convert the total islets number to Islets Equivalent of an average diameter of 150 μm (IEQ).
Suspended islets isolated from the pancreas are separated in different layers based on their degree of purity. An islet sample from each layer of the final product is then analyzed: the total number of islets and the IEQ are then calculated based on the number, the purity and the size of the islets, using standardized methods. Since in the context of the present invention the islet-like structures are obtained from cultured cells, that are adherent to the plate, the inventors assessed both the number and the diameter of the islet-like structures by analyzing 200 10× micrographs, randomly selected, per experiment, and expressed the results as %±SD (FIG. 12 A) and IEQ normalized to 100 islets (FIG. 12 C).
Thanks to the above-illustrated features, the spheroid pancreatic islet-like cell structure according to the present invention are particularly suitable for use both in clinical application, such as in a method treatment of diabetes by pancreatic islet transplantation, and in in vitro applications, such as screening methods for identifying substances capable of promoting the expression of one or more pancreatic hormones by pancreatic islet cells or for identifying substances capable of exerting a cytotoxic effect on pancreatic islet cells.
The following examples disclose in detail the differentiation of HLSCs into spheroid pancreatic islet-like cell structures according to the present invention, using protamine as the poly-cationic substance. However, the examples are not intended to limit the purpose and scope of the invention.
EXAMPLES 1. HLSC Culture and Expansion
1.1 Isolation, Characterization and Culture of HLSCs
HLSCs lines are isolated from healthy liver tissues of patients undergoing hepatectomies and characterized as previously described (Mohamad Buang M L et al. Arch Med Res. 2012; 43(1): 83-8). Specifically, HLSCs are seeded in 75 cm3 culture flasks and cultured in a medium (see Table 1) containing a 3 to 1 proportion of α-minimum essential medium and endothelial cell basal medium-1, supplemented with L-glutamine 2 mM, penicillin 100 UI/ml/streptomycin 100 μg/ml and 10% Fetal Bovine Serum. Cells are maintained in a humidified 5% CO2 incubator at 37° C.
1.2 Detachment of HLSCs
Once up to a ≈80% confluence, cells are washed twice with PBS and incubated with trypsin-EDTA 1× for about 5 minutes at 37° C., in order to induce cell detachment. Trypsin activity is subsequently neutralized by adding RPMI supplemented with L-glutamine 2 mM, penicillin 100 UI/ml/streptomycin 100 μg/ml and 10% Fetal Bovine Serum. Then, cells are harvested by centrifugation at 1200 rpm for 5 minutes, the supernatant is removed and the pellet re-suspended in culture medium and split among five 75 cm3 culture flasks.
1.3 Cryopreservation of Cells
Cells up to a ≈80% confluence are detached and harvested by centrifugation as described in paragraph 1.2. The cells are counted and 106 cells per vial are cryopreserved. The cell pellet is resuspended in a 1 ml solution containing 90% FBS and 10% dimethyl sulphoxide (DMSO) and placed into pre-cooled cryovial/s. The cryovials are frozen at −80° C. overnight before being placed into the liquid nitrogen container at −196° C.
1.4 Thawing of Cryopreserved Cells
A vial of frozen HLSCs is removed from the liquid nitrogen tank and placed in a beaker of water pre-warmed to 37° C. Once the cell suspension has completely thawed, it is placed into a sterile 50 ml falcon tube with 10 ml of sterile media and centrifuged at 1200 rpm for 5 minutes. The cell pellet is then resuspended in culture medium and split in three 75 cm3 culture flask and left to attach overnight at 37° C. in a humidified 5% CO2 incubator. The medium is changed the following day.
2. HLSC Differentiation into Islets-Like Structures
HLSCs at a density of 12×103/cm2 are seeded in 25 cm3 culture flasks or in a Petri dish 100×20 mm in the differentiation culture medium 1 (see Table 1) consisting in RPMI 1640 or DMEM supplemented with 10% Fetal Bovine Serum, glucose 11.6 mM, protamin chloride 10 μg/ml, L-glutamine 2 mM and penicillin 100 UI/ml/streptomycin 100 μg/ml. Cells are placed, for a period of 4 days, without changing the medium, in a humidified 5% CO2 incubator at 37° C. On day 5, the medium is replaced with differentiation culture medium 2 (see Table 3) consisting in RPMI 1640 or DMEM supplemented with 10% FBS, glucose 11.6 mM, L-glutamine 2 mM and penicillin 100 UI/ml/streptomycin 100 μg/ml. Medium is subsequently changed every other day. Within 2 to 4 days cells are expected to start organizing in islets-like structures that reach a maximum number after 14-18 days (FIG. 1).
TABLE 1
HLSC culture medium
Final concentration Volume (ml)
Final volume 250
α-MEM 165
EBM 55
FBS 10% 25
Penicillin 10000 UI/ml/ 100 UI/ml/100 ug/ml 2.5
streptomycin 10 mg/ml
Glutamine
200 mM 2 mM 2.5
TABLE 2
HLSC differentiation medium 1
Final concentration Volume (ml)
Final volume 250
RPMI 1640 or DMEM 216.85
FBS 10% 25
Penicillin/streptomycin 100 UI/ml/100 ug/ml 2.5
Glutamine 200 mM 2 mM 2.5
Glucose 1M 11.6 mM 2.9
Protamine 10 mg/ml 10 ug/ml 0.25
TABLE 3
HLSC differentiation medium 2
Final concentration Volume (ml)
Final volume 250
RPMI or DMEM 217.1
FBS 10% 25
Penicillin/streptomycin 100 UI/ml/100 ug/ml 2.5
Glutamine 200 mM 2 mM 2.5
Glucose 1M 11.6 mM 2.9
The results obtained are illustrated in the appended drawings, the content of which is briefly illustrated herein below.
FIG. 1: representative 20× micrographs showing HLSCs in basal culture medium (A) and following culture in differentiation medium with protamine (B-D). After 24 h, the cells changed in morphology and started forming small clusters (B) that progressively increased in both size and number, reaching a maximum number after a culture period of 18 days (C). (D) 40× micrograph showing islets-like structures after a culture period of 18 days. (E) Graph showing the total number of islets-like structures per 25 cm3 culture flask after 18 days of culture. (F) Graph representing the mean±SD number of islets-like structures after 18 days of culture (n=10).
FIGS. 2-8: representative pictures showing islet-like structures characterization by immunofluorescence after 14 days of culture: islets-like structures become positively stained for both PDX-1 and NgN3 (FIG. 2), insulin and glucagon (FIG. 3), C-peptide and GLUT-2 (FIG. 4), somatostatin (FIG. 5), ghrelin and PP (FIG. 6), collagen IV (FIG. 7) and von Willebrand Factor (FIG. 8).
FIG. 9: representative pictures showing immunofluorescence characterization of islet-like structures cells following islet-like structures disaggregation (trypsin 1×) after 14 days of culture.
FIG. 10: graph showing both blood glucose and human C-peptide levels in non-diabetic SCID mice (solid line, CTRL), streptozotocin-induced (55 mg/kg/day for 5 days) diabetic SCID mice (dotted line, DM) and streptozotocin-induced diabetic SCID mice that received HLSC derived islets-like structures implant (5000 IEQ/kg) under the renal capsule (dashed line, DM+ILS), before and 13 days following the implant. Compared to diabetic mice, diabetic mice that received islets-like structures implant had a significant decrease in blood glucose levels. This was paralleled by a concomitant increase in human C-peptide, that remained undetectable in both non diabetic SCID mice and in diabetic SCID mice that did not receive the implant.
FIG. 11: graph showing islets-like structure (ILS) formation following HLSCs culture in RPMI or DMEM based medium supplemented with FBS 10% (F), FBS 10%+ glucose 11.6 mM (FG) or FBS 10%+ glucose 11.6 mM+ protamine chloride 10 μg/ml (FGP) for 48 hours.
FIG. 12: graphs showing the size distribution (A), the mean±SD diameter (B) and IEQ/100 ILS (C) of islets-like structures derived from HLSCs following 14 days of culture in RPMI based medium supplemented with FBS 10%+ glucose 11.6 mM+ protamine chloride 10 μg/ml.
FIG. 13: Glucose does not affect islets-like structures (ILS) formation (A), but plays a key role in inducing endocrine specification and insulin/glucagon expression (B). A. Representative picture and graph showing islets-like structure formation following culture (4 days) in RPMI/DMEM supplemented with protamine chloride 10 ug/ml (P) without or with different glucose concentrations (6, 11.6 and 28 mM). B. Representative picture showing PDX-1, NgN3, insulin and glucagon expression in RPMI-based medium with glucose 1 or 25 mM.
FIG. 14: representative pictures showing the expression of PDX-1, NgN3, GLUT-2, C-peptide, glucagon and somatostatin in cells cultivated in a poly-lysine differentiation medium.

Claims (10)

The invention claimed is:
1. A method of preparing a spheroid pancreatic islet-like cell structure, comprising culturing a stem cell in a first differentiation liquid cell culture medium comprising a poly-cationic substance selected from the group consisting of poly-lysine and a soluble salt of protamine.
2. The method according to claim 1, wherein said culturing the stem cell in the first differentiation liquid cell culture medium comprising a poly-cationic substance is performed for a period of time of 2 to 4 days.
3. The method according to claim 1, wherein the poly-cationic substance is a soluble salt of protamine.
4. The method according to claim 3, wherein the soluble salt of protamine is protamine sulfate or protamine hydrochloride.
5. The method according to claim 4, wherein the protamine sulfate or protamine hydrochloride is at a concentration comprised within the range of 5 to 20 mM.
6. The method according to claim 1, wherein the poly-cationic substance is poly-lysine.
7. The method according to claim 1, which further comprises replacing the first differentiation liquid cell culture medium comprising a poly-cationic substance with a second differentiation liquid cell culture medium not comprising a poly-cationic substance and culturing until completely differentiated cells are obtained.
8. The method according to claim 7, wherein the culturing in the second differentiation liquid cell culture medium not comprising a poly-cationic substance is performed for a period of time of at least 2 days.
9. The method according to claim 1, wherein the stem cell is an adult stem cell.
10. The method according to claim 9, wherein the adult stem cell is a human liver stem cell.
US15/103,317 2013-12-16 2014-12-16 Pancreatic islet-like cell structures and a method of preparing thereof Active 2035-05-13 US10174286B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP13197368.7 2013-12-16
EP13197368 2013-12-16
EP13197368 2013-12-16
PCT/EP2014/077976 WO2015091493A1 (en) 2013-12-16 2014-12-16 Pancreatic islet-like cell structures and a method of preparing thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/077976 A-371-Of-International WO2015091493A1 (en) 2013-12-16 2014-12-16 Pancreatic islet-like cell structures and a method of preparing thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/203,869 Division US11453863B2 (en) 2013-12-16 2018-11-29 Pancreatic islet-like cell structures and a method of preparing thereof

Publications (2)

Publication Number Publication Date
US20160312189A1 US20160312189A1 (en) 2016-10-27
US10174286B2 true US10174286B2 (en) 2019-01-08

Family

ID=49765387

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/103,317 Active 2035-05-13 US10174286B2 (en) 2013-12-16 2014-12-16 Pancreatic islet-like cell structures and a method of preparing thereof
US16/203,869 Active 2036-08-14 US11453863B2 (en) 2013-12-16 2018-11-29 Pancreatic islet-like cell structures and a method of preparing thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/203,869 Active 2036-08-14 US11453863B2 (en) 2013-12-16 2018-11-29 Pancreatic islet-like cell structures and a method of preparing thereof

Country Status (11)

Country Link
US (2) US10174286B2 (en)
EP (1) EP3083941B1 (en)
JP (2) JP6643993B2 (en)
KR (1) KR102338815B1 (en)
CN (2) CN111635883A (en)
AU (1) AU2014364632B2 (en)
BR (1) BR112016013623B1 (en)
CA (1) CA2933135C (en)
DK (1) DK3083941T3 (en)
ES (1) ES2941686T3 (en)
WO (1) WO2015091493A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011252915B2 (en) 2010-05-12 2016-05-19 Prokidney Bioactive renal cells
WO2020035480A1 (en) 2018-08-16 2020-02-20 Unicyte Islet Ag Viable pancreatic islet-like cell structures and a method of preparing thereof
EP3816276A1 (en) 2019-10-31 2021-05-05 Unicyte Islet AG Viable pancreatic islet-like cell structures and a method of preparing thereof
CN113637630B (en) * 2020-05-11 2024-03-29 中国科学院分子细胞科学卓越创新中心 Islet-like cell mass, and preparation method and application thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000003001A1 (en) 1998-07-10 2000-01-20 Rhode Island Hospital Liver stem cell
WO2000043498A2 (en) 1999-01-19 2000-07-27 University Of North Carolina At Chapel Hill Human liver progenitors
WO2003033697A1 (en) 2001-10-18 2003-04-24 Ixion Biotechnology, Inc. Conversion of liver stem and progenitor cells to pancreatic functional cells
WO2003078588A2 (en) 2002-03-15 2003-09-25 University Of North Carolina At Chapel Hill Primitive and proximal hepatic stem cells
EP1394263A1 (en) 2001-05-16 2004-03-03 Kanagawa Academy Of Science And Technology Method of detecting and separating undifferentiated liver cellss using dlk
US20050074876A1 (en) 2003-10-03 2005-04-07 Inserm Bipotential liver cell lines from wild-type mammalian liver tissue
US20060148074A1 (en) * 1996-08-30 2006-07-06 Invitrogen Corporation Serum-free mammalian cell culture medium, and uses thereof
US20060246418A1 (en) 2005-04-04 2006-11-02 Montminy Marc R Method for screening compounds & uses therefor
WO2006126219A1 (en) 2005-05-26 2006-11-30 Fresenius Medical Care Deutschland G.M.B.H. Liver progenitor cells
US20090053758A1 (en) 1999-10-01 2009-02-26 University Of North Carolina At Chapel Hill Processes for clonal growth of hepatic progenitor cells
WO2009090424A1 (en) 2008-01-14 2009-07-23 University Of Brighton Cell culture system for pancreatic islands
US20100093090A1 (en) 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
CN102517248A (en) 2011-12-30 2012-06-27 中日友好医院 In-vitro induced pancreas-islet-like structure forming method

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU631112B2 (en) * 1988-01-11 1992-11-19 Amylin Corporation Treatment of type 2 diabetes mellitus
IL149933A0 (en) * 1999-12-06 2002-11-10 Gen Hospital Corp Pancreatic stem cells and their use in transplantation
EP1264879A1 (en) * 2001-06-08 2002-12-11 Leadd B.V. Isolation of a cluster of cells from an isolated organ using magnetic particle
EP1463798A4 (en) * 2001-12-07 2005-01-19 Geron Corp Islet cells from human embryonic stem cells
US20030162290A1 (en) * 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
CN1536075A (en) * 2003-04-09 2004-10-13 中国人民解放军军事医学科学院野战输 Method for inducing bone marrow mesenchymal stem cells to differentiate into insulin-like cells
CA2531312A1 (en) * 2003-07-29 2005-02-17 Unilever Plc Food product comprising phytosterols
US7507581B2 (en) * 2005-04-05 2009-03-24 Synthecon, Inc. Inhibition of pancreatic islet aggregation
WO2007039986A1 (en) * 2005-10-05 2007-04-12 Osaka University Method of obtaining pancreatic endocrine cells from adipose tissue-origin cells
CN101356264B (en) * 2005-12-21 2014-05-14 鲁汶大学 Isolated liver stem cells
US7716503B2 (en) 2006-12-14 2010-05-11 Inventec Corporation Extension card incorporating power management device
JP5196522B2 (en) * 2007-06-27 2013-05-15 国立大学法人東北大学 Evaluation method of islets for transplantation
CN101896188B (en) * 2007-10-15 2013-01-02 弗雷森纽斯医疗护理德国有限责任公司 Use of microvesicles (MVS) for preparing a medicament having adjuvant activity on endothelial cell transplantation, particularly in the treatment of diabetes by pancreatic islet transplantation, and related method
US20110008301A1 (en) * 2008-03-15 2011-01-13 Trivedi H L Human adipose derived insulin making mesenchymal stem cells for treating diabetes mellitus
CN101603027A (en) * 2008-06-12 2009-12-16 西北农林科技大学 External evoked human marrow mesenchymal stem cell is divided into the method for insulin-like cell
EP3441458B9 (en) * 2009-02-03 2023-08-23 Koninklijke Nederlandse Akademie van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
CN102433300A (en) * 2011-11-25 2012-05-02 厚朴生物科技(苏州)有限公司 Method for constructing pancreatic stem cell line from human insulin and differentiating to insulin secretion cell

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060148074A1 (en) * 1996-08-30 2006-07-06 Invitrogen Corporation Serum-free mammalian cell culture medium, and uses thereof
WO2000003001A1 (en) 1998-07-10 2000-01-20 Rhode Island Hospital Liver stem cell
WO2000043498A2 (en) 1999-01-19 2000-07-27 University Of North Carolina At Chapel Hill Human liver progenitors
US20090053758A1 (en) 1999-10-01 2009-02-26 University Of North Carolina At Chapel Hill Processes for clonal growth of hepatic progenitor cells
EP1394263A1 (en) 2001-05-16 2004-03-03 Kanagawa Academy Of Science And Technology Method of detecting and separating undifferentiated liver cellss using dlk
WO2003033697A1 (en) 2001-10-18 2003-04-24 Ixion Biotechnology, Inc. Conversion of liver stem and progenitor cells to pancreatic functional cells
US20030138951A1 (en) 2001-10-18 2003-07-24 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
WO2003078588A2 (en) 2002-03-15 2003-09-25 University Of North Carolina At Chapel Hill Primitive and proximal hepatic stem cells
US20050074876A1 (en) 2003-10-03 2005-04-07 Inserm Bipotential liver cell lines from wild-type mammalian liver tissue
US20060246418A1 (en) 2005-04-04 2006-11-02 Montminy Marc R Method for screening compounds & uses therefor
WO2006126219A1 (en) 2005-05-26 2006-11-30 Fresenius Medical Care Deutschland G.M.B.H. Liver progenitor cells
WO2006126236A1 (en) 2005-05-26 2006-11-30 Fresenius Medical Care Deutschland G.M.B.H. Liver progenitor cells
WO2009090424A1 (en) 2008-01-14 2009-07-23 University Of Brighton Cell culture system for pancreatic islands
US20100093090A1 (en) 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
CN102517248A (en) 2011-12-30 2012-06-27 中日友好医院 In-vitro induced pancreas-islet-like structure forming method

Non-Patent Citations (30)

* Cited by examiner, † Cited by third party
Title
Bose et al. Human embryonic stem cell differentiation into insulin secreting β-cells for diabetes. Cell Biology International, (2012) 36, 1013-1020.
Buang et al, In vitro generation of functional insulin-producing cells from lipoaspirated human adipose tissue-derived stem cells. Archives of Medical Research 43 (2012) 83-88.
Carnevale et al. In vitro differentiation into insulin-producing β-cells of stem cells isolated from human amniotic fluid and dental pulp. Digestive and Liver Disease 45 (2013) 669-676.
Chandra et al. Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLOS One, vol. 6, issue 6, Jun. 2011, pp. 1-12.
Chandra et al., Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLOS One, vol. 6, No. 6 (Jun. 2011) pp. 1-12. *
Chun et al. Adhesive growth of pancreatic islet cells on a polyglycolic acid fibrous scaffold. Transplantation Proceedings, vol. 40, No. 5, Jun. 1, 2008, pp. 1658-1663.
Duan et al. Improvement in the methods for separation and culture of neural stem cells in striatum of embryonic rats Chines J Clin. vol. 9, No. 26, Jul. 14, 2005, pp. 92-93.
Fischer et al., In vitro cytotoxicity testing of polycations: influence of polymer structure on cell viability and hemolysis. Biomaterials, vol. 24, No. 7 (Mar. 2003) pp. 1121-1131. *
Forbes et al. Hepatic Stem Cells, Journal of Pathology, vol. 197, No. 4, pp. 510-518, May 30, 2002.
Herrera et al. Isolation and characterization of a stem cell population from adult human liver. Stem Cells, vol. 24, No. 12, Jan. 1, 2006, pp. 2840-2850.
Hillen et al. Analysis of tet Operator-TET Repressor Complexes by Thermal Denaturation Studies, Nucleic Acid Research, vol. 10, No. 19, pp. 6085-6097, 1982.
Hillen et al. Control of Expression of the Tn10-encoded Tetracycline Resistance Genes, Journal of Mol. Biology, vol. 169, pp. 707-721, 1983.
Hillen et al. Mechanisms Underlying Expression of Tn10 Ecnoded Tetracylcine Resistance, Microbiology, pp. 345-369, 1994.
Jamal et al. Morphogenetic plasticity of adult human pancreatic islets of Langerhans. Cell Death and Differentiation, vol. 12, No. 7, Apr. 8, 2005, pp. 702-712.
Kroon et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insuline-secreting cells in vivo. Nature Biotechnology, vol. 26, No. 4, Apr. 2008, pp. 443-452.
Lumelsky et al. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic Islets. Science vol. 292, No. 5520, May 18, 2001, pp. 1389-1394.
Malhi et al. Isolation of Human Progenitor Liver Epithellal Cells with Extensive Replication Capacity and Differentiation into Mature Hepatocytes. Journal of Cell Science, vol. 115, No. 13, pp. 2679-2688, Jul. 1, 2002.
Milanesi et al. Differentiation of nestin-positive cells derived from bone marrow into pancreatic endocrine and ductal cells in vitro. Journal of Endocrinology (2011) 209, 193-201.
Mitaka et al. Reconstruction of Hepatic Organoid by Hepatic Stem Cells, Journal of HBP Surgery, pp. 697-703, 2002.
Petersen et al. Hepatic Oval Cells Express Stem Cell Markers Thy-1 in the Rat, Heptalogy, vol. 27, No. 2, pp. 433-445, Feb. 1998.
Petersen et al. Mouse A6-Positive Hepatic Oval Cells Also Express Several Hematopoietic Stem Cell Markers, Heptalogy, vol. 37, No. 3, pp. 632-640, Mar. 2003.
Prabakar et al. Generation of glucose-responsive, insuline-producing cells from human umbilical cord blood-derived mesenchynal stem cells. Cell Transplantation, vol. 21, pp. 1321-1339, 2012.
Ramiya et al. Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nature Medicine, vol. 6, No. 3, Mar. 1, 2000, pp. 278-282.
Rezania et al. Maturation of human embryonic stern cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice, Diabetes, vol. 61, Aug. 2012, pp. 2016-2029.
Schwartz et al. Multipotent Adult Progenitor Cells from Bone Marrow Differentiate into Functional Hepatocyte-like Cells. Journal of Clinical Investigation, vol. 109, No. 10, pp. 1291-1302, May 2002.
Suzuki et al. Clonal Identification and Characterization of Self-Renewing Pluripotent Stem Cells in the Developing Liver, Journal of Cell Biology, vol. 156, No. 1, pp. 173-184, Jan. 7, 2002.
Suzuki et al. Flow Cytometric Separation and Enrichment of Hepatic Progenitor Cells in the Developing Mouse Liver. Hepatology, vol. 32, No. 6, pp. 1230-1239, Dec. 6, 2000.
Wen et al. A spheroid-based 3D culture model for pancreatic cancer drug testing, using the acid phosphatase assay. Brazilian Journal of Medical and Biological Research, vol. 46, No. 7, Aug. 1, 2013, pp. 634-642.
Yang et al. In vitro trans-differentiation of adult hepatic stem cells into pancreatic andocrine hormone-producing cells. PNAS USA, 99, 8078-8083, Jun. 11, 2002.
Yang et al. In Vitro Transdifferentiation of adult hepatic stem cells into pancreatic endocrine cells. Blood. vol. 98, No. 11, part 01, Nov. 16. 2001, pp. 548A.

Also Published As

Publication number Publication date
DK3083941T3 (en) 2023-03-13
JP6643993B2 (en) 2020-02-12
CN105814193A (en) 2016-07-27
CN105814193B (en) 2020-05-08
CA2933135C (en) 2023-08-01
EP3083941B1 (en) 2023-02-22
US20160312189A1 (en) 2016-10-27
WO2015091493A1 (en) 2015-06-25
AU2014364632B2 (en) 2021-04-01
BR112016013623B1 (en) 2023-03-21
AU2014364632A1 (en) 2016-06-16
CN111635883A (en) 2020-09-08
US11453863B2 (en) 2022-09-27
JP2020014477A (en) 2020-01-30
KR102338815B1 (en) 2021-12-13
BR112016013623A2 (en) 2017-08-08
JP2017500859A (en) 2017-01-12
EP3083941A1 (en) 2016-10-26
CA2933135A1 (en) 2015-06-25
US20190119648A1 (en) 2019-04-25
KR20160098439A (en) 2016-08-18
ES2941686T3 (en) 2023-05-24

Similar Documents

Publication Publication Date Title
Shahjalal et al. Generation of pancreatic β cells for treatment of diabetes: advances and challenges
US11453863B2 (en) Pancreatic islet-like cell structures and a method of preparing thereof
ES2967942T3 (en) Generation of functional beta cells from endocrine progenitors derived from human pluripotent stem cells
Chandra et al. Generation of pancreatic hormone-expressing islet-like cell aggregates from murine adipose tissue-derived stem cells
Cheng et al. Human adipose-derived stem cells: Isolation, characterization and current application in regeneration medicine
WO2011101834A1 (en) A method for obtaining mesenchymal stem cells, media, methods and composition thereof
JP5570814B2 (en) Muscle-derived cells for the treatment of gastroesophageal pathology and their preparation and use
Cañibano-Hernández et al. Hyaluronic acid promotes differentiation of mesenchymal stem cells from different sources toward pancreatic progenitors within three-dimensional alginate matrixes
US20240124843A1 (en) Functional feline pancreatic cells from adipose tissue
Weizman et al. The effect of endothelial cells on hESC-derived pancreatic progenitors in a 3D environment
WO2020035480A1 (en) Viable pancreatic islet-like cell structures and a method of preparing thereof
Zweigerdt The art of cobbling a running pump—will human embryonic stem cells mend broken hearts?
CN110923192B (en) Long-term in vitro culture method of mature hepatocytes
Anzalone et al. Mesenchymal Stromal Cells From Wharton's Jelly (WJ-MSCs): Coupling Their Hidden Differentiative Program to Their Frank Immunomodulatory Phenotype
US20110014260A1 (en) System and method for liver cell culture and maturation
Li et al. Pancreatic line cell differentiation of bone marrow mesenchymal stromal cells in acellular pancreatic scaffolds
Luo et al. The efficiency of stem cell differentiation into functional beta cells for treating insulin-requiring diabetes: Recent advances and current challenges
Shi et al. Advancements in culture technology of adipose-derived stromal/stem cells: implications for diabetes and its complications
Bose et al. Cell Therapy for Diabetes
WO2021083962A1 (en) Viable pancreatic islet-like cell structures and a method of preparing thereof
Li et al. Pancreatic line Cell Differentiation of bone marrow Mesenchymal Stromal Cells in Acellular Pancreatic Scaffolds Running title: BMSCs Differentiation in APB
Omer et al. Human embryo pancreatic stem cells differentiating into active insulin secreting islet-like structure
Domínguez-Bendala et al. Adult Stem Cells and Pancreatic Differentiation

Legal Events

Date Code Title Description
AS Assignment

Owner name: FRESENIUS MEDICAL CARE DEUTSCHLAND GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TETTA, CIRO;CAMUSSI, GIOVANNI;GIUNTI, SARA;AND OTHERS;SIGNING DATES FROM 20160510 TO 20160513;REEL/FRAME:038871/0012

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4